1
|
Carducci M, Massai L, Lari E, Semplici B, Aruta MG, De Simone D, Piu P, Montomoli E, Berlanda Scorza F, Grappi S, Iturriza-Gómara M, Canals R, Rondini S, Rossi O. Qualification of an enzyme-linked immunosorbent assay for quantification of anti-Vi IgG in human sera. Front Immunol 2024; 15:1466869. [PMID: 39478859 PMCID: PMC11521798 DOI: 10.3389/fimmu.2024.1466869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/20/2024] [Indexed: 11/02/2024] Open
Abstract
Effective vaccines against Salmonella Typhi, targeting the Vi capsular polysaccharide, have been developed and are being introduced into routine immunization in endemic countries. Vi conjugated vaccines are also being tested in new multi-component vaccine formulations. Simple, high-throughput and cost-effective assays to quantify Vi-specific IgG in clinical sera are needed. In this study we present the development and qualification of a new anti-Vi ELISA with continuous readout, which expresses results as ELISA Unit/mL (EU/mL). We have qualified the assay in terms of precision, linearity and specificity, demonstrating performance in line with a commercially available anti-Vi ELISA. We have also calibrated the assay against the 16/138 anti-Vi international standard and established conversion factor between EU/mL and international units/mL, to allow comparability of results across studies. In summary, this new assay met all the suitability criteria and is being used to evaluate anti-Vi responses in clinical studies.
Collapse
Affiliation(s)
- Martina Carducci
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Luisa Massai
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | | | | | | | - Daniele De Simone
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | | | - Emanuele Montomoli
- VisMederi S.r.l., Siena, Italy
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | | | | | | | - Rocio Canals
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Simona Rondini
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Omar Rossi
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| |
Collapse
|
2
|
Agarwal N, Gupta N, Nishant, H S S, Dutta T, Mahajan M. Typhoid Conjugate Vaccine: A Boon for Endemic Regions. Cureus 2024; 16:e56454. [PMID: 38650789 PMCID: PMC11034893 DOI: 10.7759/cureus.56454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 04/25/2024] Open
Abstract
Typhoid fever has the highest disease burden in countries in low- and middle-income countries, primarily located in Asia and Sub-Saharan Africa. Previous typhoid vaccines such as the live attenuated typhoid (Ty21a) vaccine and Vi (virulence) capsular polysaccharide vaccine had the limitation that they could not be administered with other standard childhood immunizations and were ineffective in children under two years of age. To address these shortcomings of the previous vaccines, typhoid conjugate vaccines (TCVs) were developed and prequalified by the World Health Organization. Cross-reacting material and tetanus toxoid are widely used as carrier proteins in TCVs. According to various studies, TCV has higher efficacy, has a more extended protection period, and is safe and immunogenic in infants as young as six months. This review article aims to comprehensively appraise the data available on TCVs' efficacy, duration of protection, safety, and immunogenicity in endemic regions.
Collapse
Affiliation(s)
- Nitesh Agarwal
- Department of Pediatrics, Southern Gem Hospital, Hyderabad, IND
| | - Naveen Gupta
- Department of Pediatrics, Happy Family Hospital, Karnal, IND
| | - Nishant
- Department of Pediatrics, Nihan Medical Children Hospital, Patna, IND
| | - Surendra H S
- Department of Pediatrics, Natus Women and Children Hospital, Bengaluru, IND
| | - Trayambak Dutta
- Department of Infectious Disease, Zydus Lifesciences, Ahmedabad, IND
| | | |
Collapse
|
3
|
Jossi SE, Arcuri M, Alshayea A, Persaud RR, Marcial-Juárez E, Palmieri E, Di Benedetto R, Pérez-Toledo M, Pillaye J, Channell WM, Schager AE, Lamerton RE, Cook CN, Goodall M, Haneda T, Bäumler AJ, Jackson-Jones LH, Toellner KM, MacLennan CA, Henderson IR, Micoli F, Cunningham AF. Vi polysaccharide and conjugated vaccines afford similar early, IgM or IgG-independent control of infection but boosting with conjugated Vi vaccines sustains the efficacy of immune responses. Front Immunol 2023; 14:1139329. [PMID: 37033932 PMCID: PMC10076549 DOI: 10.3389/fimmu.2023.1139329] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/13/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction Vaccination with Vi capsular polysaccharide (Vi-PS) or protein-Vi typhoid conjugate vaccine (TCV) can protect adults against Salmonella Typhi infections. TCVs offer better protection than Vi-PS in infants and may offer better protection in adults. Potential reasons for why TCV may be superior in adults are not fully understood. Methods and results Here, we immunized wild-type (WT) mice and mice deficient in IgG or IgM with Vi-PS or TCVs (Vi conjugated to tetanus toxoid or CRM197) for up to seven months, with and without subsequent challenge with Vi-expressing Salmonella Typhimurium. Unexpectedly, IgM or IgG alone were similarly able to reduce bacterial burdens in tissues, and this was observed in response to conjugated or unconjugated Vi vaccines and was independent of antibody being of high affinity. Only in the longer-term after immunization (>5 months) were differences observed in tissue bacterial burdens of mice immunized with Vi-PS or TCV. These differences related to the maintenance of antibody responses at higher levels in mice boosted with TCV, with the rate of fall in IgG titres induced to Vi-PS being greater than for TCV. Discussion Therefore, Vi-specific IgM or IgG are independently capable of protecting from infection and any superior protection from vaccination with TCV in adults may relate to responses being able to persist better rather than from differences in the antibody isotypes induced. These findings suggest that enhancing our understanding of how responses to vaccines are maintained may inform on how to maximize protection afforded by conjugate vaccines against encapsulated pathogens such as S. Typhi.
Collapse
Affiliation(s)
- Siân E. Jossi
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Melissa Arcuri
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- GSK Vaccines Institute for Global Health SRL, Siena, Italy
| | - Areej Alshayea
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Ruby R. Persaud
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Edith Marcial-Juárez
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Elena Palmieri
- GSK Vaccines Institute for Global Health SRL, Siena, Italy
| | | | - Marisol Pérez-Toledo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Jamie Pillaye
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Will M. Channell
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Anna E. Schager
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Rachel E. Lamerton
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Charlotte N. Cook
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Margaret Goodall
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Takeshi Haneda
- Laboratory of Microbiology, School of Pharmacy, Kitasato University, Tokyo, Japan
| | - Andreas J. Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA, United States
| | - Lucy H. Jackson-Jones
- Division of Biomedical and Life Sciences, Lancaster University, Lancaster, United Kingdom
| | - Kai-Michael Toellner
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Calman A. MacLennan
- Bill & Melinda Gates Foundation, London, United Kingdom
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ian R. Henderson
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | | | - Adam F. Cunningham
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
4
|
Berlanda Scorza F, Martin LB, Podda A, Rappuoli R. A strategic model for developing vaccines against neglected diseases: An example of industry collaboration for sustainable development. Hum Vaccin Immunother 2022; 18:2136451. [PMID: 36495000 PMCID: PMC9746511 DOI: 10.1080/21645515.2022.2136451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 09/30/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
Infectious diseases continue to disproportionately affect low- and middle-income countries (LMICs) and children aged <5 y. Developing vaccines against diseases endemic in LMICs relies mainly on strong public-private collaborations, but several challenges remain. We review the operating model of the GSK Vaccines Institute for Global Health (GVGH), which aims to address these challenges. The model involves i) selection of vaccine targets based on priority ranking for impact on global health; ii) development from design to clinical proof-of-concept; iii) transfer to an industrial partner, for further technical/clinical development, licensing, manufacturing, and distribution. Cost and risks associated with pre-clinical and early clinical development are assumed by GVGH, increasing the probability to make the vaccine more affordable in LMICs. A conjugate vaccine against typhoid fever, Vi-CRM197, has recently obtained WHO prequalification, within a year from licensure in India, demonstrating the success of the GVGH model for development and delivery of global health vaccines.
Collapse
Affiliation(s)
| | | | - Audino Podda
- GSK Vaccines Institute for Global Health, Siena, Italy
| | - Rino Rappuoli
- GSK Vaccines Institute for Global Health, Siena, Italy
- GSK, Siena, Italy
| |
Collapse
|
5
|
Thuluva S, Paradkar V, Matur R, Turaga K, Gv SR. A multicenter, single-blind, randomized, phase-2/3 study to evaluate immunogenicity and safety of a single intramuscular dose of biological E's Vi-capsular polysaccharide-CRM 197 conjugate typhoid vaccine (TyphiBEV TM) in healthy infants, children, and adults in comparison with a licensed comparator. Hum Vaccin Immunother 2022; 18:2043103. [PMID: 35333702 PMCID: PMC9196756 DOI: 10.1080/21645515.2022.2043103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The current scenario of typhoid fever warrants early prevention with typhoid conjugate vaccines in susceptible populations to provide lifelong protection. We conducted a multicenter, single-blind, randomized, Phase 2/3 study to assess the immunogenicity and safety of Biological E’s Typhoid Vi-CRM197 conjugate vaccine (TyphiBEVTM) compared to Vi-TT conjugate vaccine manufactured by Bharat Biotech International Limited (Typbar-TCV; licensed comparator) in healthy infants, children, and adults from India. The study’s primary objective was to assess the non-inferiority of TyphiBEVTM in terms of the difference in the proportion of subjects seroconverted with a seroconversion threshold value of ≥2.0 µg/mL against Typbar-TCV. A total of 622 healthy subjects (311 each in both vaccine groups) were randomized and received the single dose of the study vaccine. The TyphiBEVTM group demonstrated noninferiority compared to the Typbar-TCV group at Day 42. The lower 2-sided 95% confidence interval limit of the group difference was −.34%, which met the non-inferiority criteria of ≥10.0%. The geometric mean concentration (24.79 µg/mL vs. 26.58 µg/mL) and proportion of subjects who achieved ≥4-fold increase in antiVi IgG antibody concentrations (96.95% vs. 97.64%) at Day 42 were comparable between the TyphiBEVTM and Typbar-TCV vaccine groups. No apparent difference was observed in the safety profile between both vaccine groups. All adverse events reported were mild or moderate in intensity in all age subsets. This data demonstrates that TyphiBEVTM is non-inferior to TypbarTCV in terms of immunogenicity, and the overall safety and reactogenicity in healthy infants, children, and adults studied from India was comparable.
Collapse
Affiliation(s)
- Subhash Thuluva
- Clinical Development Department, Biological E Limited, Hyderabad, India
| | - Vikram Paradkar
- Clinical Development Department, Biological E Limited, Hyderabad, India
| | - Ramesh Matur
- Clinical Development Department, Biological E Limited, Hyderabad, India
| | - Kishore Turaga
- Clinical Development Department, Biological E Limited, Hyderabad, India
| | - Subba Reddy Gv
- Clinical Development Department, Biological E Limited, Hyderabad, India
| |
Collapse
|
6
|
Stefanetti G, MacLennan CA, Micoli F. Impact and Control of Sugar Size in Glycoconjugate Vaccines. Molecules 2022; 27:molecules27196432. [PMID: 36234967 PMCID: PMC9572008 DOI: 10.3390/molecules27196432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 11/17/2022] Open
Abstract
Glycoconjugate vaccines have contributed enormously to reducing and controlling encapsulated bacterial infections for over thirty years. Glycoconjugate vaccines are based on a carbohydrate antigen that is covalently linked to a carrier protein; this is necessary to cause T cell responses for optimal immunogenicity, and to protect young children. Many interdependent parameters affect the immunogenicity of glycoconjugate vaccines, including the size of the saccharide antigen. Here, we examine and discuss the impact of glycan chain length on the efficacy of glycoconjugate vaccines and report the methods employed to size polysaccharide antigens, while highlighting the underlying reaction mechanisms. A better understanding of the impact of key parameters on the immunogenicity of glycoconjugates is critical to developing a new generation of highly effective vaccines.
Collapse
Affiliation(s)
- Giuseppe Stefanetti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
- Correspondence:
| | - Calman Alexander MacLennan
- Enteric and Diarrheal Diseases, Global Health, Bill & Melinda Gates Foundation, 500 5th Ave. N, Seattle, WA 98109, USA
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
- The Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | | |
Collapse
|
7
|
Cross reacting material (CRM197) as a carrier protein for carbohydrate conjugate vaccines targeted at bacterial and fungal pathogens. Int J Biol Macromol 2022; 218:775-798. [PMID: 35872318 DOI: 10.1016/j.ijbiomac.2022.07.137] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/14/2022] [Accepted: 07/18/2022] [Indexed: 11/22/2022]
Abstract
This paper gives an overview of conjugate glycovaccines which contain recombinant diphtheria toxoid CRM197 as a carrier protein. A special focus is given to synthetic methods used for preparation of neoglycoconjugates of CRM197 with oligosaccharide epitopes of cell surface carbohydrates of pathogenic bacteria and fungi. Syntheses of commercial vaccines and laboratory specimen on the basis of CRM197 are outlined briefly.
Collapse
|
8
|
Aw R, Ashik MR, Islam AAZM, Khan I, Mainuddin M, Islam MA, Ahasan MM, Polizzi KM. Production and purification of an active CRM197 in Pichia pastoris and its immunological characterization using a Vi-typhoid antigen vaccine. Vaccine 2021; 39:7379-7386. [PMID: 34774362 DOI: 10.1016/j.vaccine.2021.10.083] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 10/06/2021] [Accepted: 10/31/2021] [Indexed: 10/19/2022]
Abstract
CRM197 is a commonly used glycoconjugate carrier that improves the immunogenicity of vaccines, particularly in infants. Despite the advantages of this diphtheria toxoid mutant, low yields, production in inclusion bodies, and the requirement for specific growth conditions have limited the breadth of successful recombinant protein expression platforms available for its expression. We evaluated Pichia pastoris as a production host, using the methanol inducible AOX1 promoter and a modified α-mating factor signal peptide for secretion into the supernatant. Final purified yields >100 mg L-1 culture were achieved when produced in a bioreactor, which is equivalent to the productivity obtained from bioprocesses using the native Corynebacterium diphtheriae host. Recombinant CRM197 was purified to ≥95% homogeneity and showed the expected endonuclease activity. Furthermore, mice immunized with a Salmonella enterica serovar Typhi capsular Vi antigen conjugated to our recombinant CRM197 showed greater than 5-fold increase in immune response. Overall, the results demonstrate that Pichia pastoris is a suitable expression host for the production of high quality CRM197 for vaccine applications.
Collapse
Affiliation(s)
- Rochelle Aw
- Department of Chemical Engineering, Imperial College London, London SW7 2AZ, UK; Imperial College Centre for Synthetic Biology, Imperial College London, SW7 2AZ, UK
| | | | | | - Imran Khan
- Incepta Vaccine Ltd, Savar, Dhaka 1341, Bangladesh
| | | | | | | | - Karen M Polizzi
- Department of Chemical Engineering, Imperial College London, London SW7 2AZ, UK; Imperial College Centre for Synthetic Biology, Imperial College London, SW7 2AZ, UK.
| |
Collapse
|
9
|
Rotavirus spike protein ΔVP8* as a novel carrier protein for conjugate vaccine platform with demonstrated antigenic potential for use as bivalent vaccine. Sci Rep 2021; 11:22037. [PMID: 34764353 PMCID: PMC8586335 DOI: 10.1038/s41598-021-01549-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/29/2021] [Indexed: 11/18/2022] Open
Abstract
Conjugate vaccine platform is a promising strategy to overcome the poor immunogenicity of bacterial polysaccharide antigens in infants and children. A carrier protein in conjugate vaccines works not only as an immune stimulator to polysaccharide, but also as an immunogen; with the latter generally not considered as a measured outcome in real world. Here, we probed the potential of a conjugate vaccine platform to induce enhanced immunogenicity of a truncated rotavirus spike protein ΔVP8*. ΔVP8* was covalently conjugated to Vi capsular polysaccharide (Vi) of Salmonella Typhi to develop a bivalent vaccine, termed Vi-ΔVP8*. Our results demonstrated that the Vi-ΔVP8* vaccine can induce specific immune responses against both antigens in immunized mice. The conjugate vaccine elicits high antibody titers and functional antibodies against S. Typhi and Rotavirus (RV) when compared to immunization with a single antigen. Together, these results indicate that Vi-ΔVP8* is a potent and immunogenic vaccine candidate, thus strengthening the potential of conjugate vaccine platform with enhanced immune responses to carrier protein, including ΔVP8*.
Collapse
|
10
|
Choi SK, Baik YO, Kim CW, Kim SK, Oh IN, Yoon H, Yu D, Lee C. An open-label, comparative, single dose, clinical Phase Ⅰ study to assess the safety and immunogenicity of typhoid conjugate vaccine (Vi-CRM197) in healthy Filipino adults. Vaccine 2021; 39:2620-2627. [PMID: 33849723 DOI: 10.1016/j.vaccine.2021.03.089] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/16/2021] [Accepted: 03/29/2021] [Indexed: 11/25/2022]
Abstract
An injectable typhoid conjugate vaccine (TCV) provides longer-lasting protection, requires fewer doses, and is suitable for children aged >2 years. In addition, TCV is preferred at most ages owing to its improved immunological properties as it overcomes the limitation of Vi polysaccharide vaccines. Here, we assessed the safety, tolerability, and immunogenicity of a TCV, Vi-CRM197, termed EuTCV, in an open-label clinical phase I study in healthy Filipino adults. This study was conducted in 75 healthy adults aged 18-45 years who were randomized in a 1:1:1 ratio based on the vaccines administered: EuTCV (Test), Typbar-TCV® (WHO prequalified vaccine) and Typhim Vi® (Vi polysaccharide vaccine). The study vaccines were administered at a dose of 25 µg of Vi-CRM197 conjugate by intramuscular injection as a single dose to each of the 25 participants/group, and their immunogenicity and overall safety were assessed for 42 days post-vaccination. All study participants (n = 25/group) completed the trial without dropouts. There were no deaths, SAEs, or events leading to premature withdrawal from the study. Anti-Vi IgG antibody titer (geometric mean titer) of EuTCV group on day 42 was 65.325 [95% CI (36.860, 115.771)], which was significantly higher than that of the WHO prequalified TCV [24.795, 95% CI (16.164, 38.033) p = 0.0055] and the Vi polysaccharide vaccine [7.998, 95% CI (3.800, 16.835) p < 0.0001]. Moreover, the seroconversion rate of EuTCV and Typbar-TCV® was 100%, but that of Typhim Vi® was only 84%. The IgG1-3 subclass titers and serum bactericidal assay results in the EuTCV group showed higher and better bactericidal capacity than the other groups. EuTCV was well tolerated and exhibited an acceptable safety profile in the study population. The Vi-CRM197 conjugate dose of 25 μg may be considered effective in terms of efficacy and safety. ClinicalTrials.gov registration number: NCT03956524.
Collapse
Affiliation(s)
- Seuk Keun Choi
- R&D Center, EuBiologics Co., Ltd., Chuncheon, Republic of Korea; Department of Biotechnology, Korea University, Seoul, Republic of Korea
| | - Yeong Ok Baik
- R&D Center, EuBiologics Co., Ltd., Chuncheon, Republic of Korea
| | - Chan Wha Kim
- Department of Biotechnology, Korea University, Seoul, Republic of Korea
| | - Soo Kyung Kim
- R&D Center, EuBiologics Co., Ltd., Chuncheon, Republic of Korea
| | - Il Nam Oh
- R&D Center, EuBiologics Co., Ltd., Chuncheon, Republic of Korea
| | - Hyeseon Yoon
- R&D Center, EuBiologics Co., Ltd., Chuncheon, Republic of Korea
| | - Dajung Yu
- R&D Center, EuBiologics Co., Ltd., Chuncheon, Republic of Korea
| | - Chankyu Lee
- R&D Center, EuBiologics Co., Ltd., Chuncheon, Republic of Korea.
| |
Collapse
|
11
|
Vashishtha VM, Kalra A. The need & the issues related to new-generation typhoid conjugate vaccines in India. Indian J Med Res 2021; 151:22-34. [PMID: 32134011 PMCID: PMC7055164 DOI: 10.4103/ijmr.ijmr_1890_17] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The low- and middle-income countries bear the highest burden of typhoid fever in the world. India, along with other South Asian countries, has a significant incidence of typhoid fever among young children though there is a paucity of published data on community burden. In spite of the availability of Vi-polysaccharide (Vi-PS) and conjugated Vi-PS vaccines, these are not adequately utilized in India and in the neighbouring countries. To address many shortcomings of the unconjugated Vi-PS vaccines, typhoid conjugate vaccines (TCVs) are developed by conjugating Vi-PS with different carrier proteins. Three such vaccines using tetanus toxoid as a carrier protein are already licensed in India. Several other Vi-PS conjugates are currently in various stages of development. The current review provides an update on the existing and upcoming new TCVs along with a detailed discussion on the various issues involved with their clinical use and limitations.
Collapse
Affiliation(s)
| | - Ajay Kalra
- Department of Pediatrics, Sarojini Naidu Medical College, Agra, Uttar Pradesh, India
| |
Collapse
|
12
|
Short Vi-polysaccharide abrogates T-independent immune response and hyporesponsiveness elicited by long Vi-CRM 197 conjugate vaccine. Proc Natl Acad Sci U S A 2020; 117:24443-24449. [PMID: 32900928 PMCID: PMC7533886 DOI: 10.1073/pnas.2005857117] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Our results suggest a rational way of designing and developing an improved typhoid conjugate vaccine and, by extension, to conjugate vaccines in general: first, modify a T-independent polysaccharide so that it no longer induces a T-independent response, then conjugate the polysaccharide to a suitable carrier protein restoring immunogenicity, thus creating a pure T-dependent antigen that induces a strongly boostable and long-lived response at an early age. Polysaccharide-protein conjugates have been developed to overcome the T-independent response, hyporesponsiveness to repeated vaccination, and poor immunogenicity in infants of polysaccharides. To address the impact of polysaccharide length, typhoid conjugates made with short- and long-chain fractions of Vi polysaccharide with average sizes of 9.5, 22.8, 42.7, 82.0, and 165 kDa were compared. Long-chain-conjugated Vi (165 kDa) induced a response in both wild-type and T cell-deficient mice, suggesting that it maintains a T-independent response. In marked contrast, short-chain Vi (9.5 to 42.7 kDa) conjugates induced a response in wild-type mice but not in T cell-deficient mice, suggesting that the response is dependent on T cell help. Mechanistically, this was explained in neonatal mice, in which long-chain, but not short-chain, Vi conjugate induced late apoptosis of Vi-specific B cells in spleen and early depletion of Vi-specific B cells in bone marrow, resulting in hyporesponsiveness and lack of long-term persistence of Vi-specific IgG in serum and IgG+ antibody-secreting cells in bone marrow. We conclude that while conjugation of long-chain Vi generates T-dependent antigens, the conjugates also retain T-independent properties, leading to detrimental effects on immune responses. The data reported here may explain some inconsistencies observed in clinical trials and help guide the design of effective conjugate vaccines.
Collapse
|
13
|
Immunogenicity and Safety of Typhoid Conjugate Vaccine in Healthy Indian Subjects: A Randomized, Active-controlled, Comparative Clinical Trial. Indian Pediatr 2020. [DOI: 10.1007/s13312-020-1890-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
14
|
Syed KA, Saluja T, Cho H, Hsiao A, Shaikh H, Wartel TA, Mogasale V, Lynch J, Kim JH, Excler JL, Sahastrabuddhe S. Review on the Recent Advances on Typhoid Vaccine Development and Challenges Ahead. Clin Infect Dis 2020; 71:S141-S150. [PMID: 32725225 PMCID: PMC7388714 DOI: 10.1093/cid/ciaa504] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Control of Salmonella enterica serovar typhi (S. typhi), the agent of typhoid fever, continues to be a challenge in many low- and middle-income countries. The major transmission route of S. typhi is fecal-oral, through contaminated food and water; thus, the ultimate measures for typhoid fever prevention and control include the provision of safe water, improved sanitation, and hygiene. Considering the increasing evidence of the global burden of typhoid, particularly among young children, and the long-term horizon for sustained, effective water and sanitation improvements in low-income settings, a growing consensus is to emphasize preventive vaccination. This review provides an overview of the licensed typhoid vaccines and vaccine candidates under development, and the challenges ahead for introduction.
Collapse
Affiliation(s)
- Khalid Ali Syed
- MSD-Wellcome Trust Hilleman Laboratories Pvt Ltd, New Delhi, India
| | - Tarun Saluja
- International Vaccine Institute, Seoul, Republic of Korea
| | - Heeyoun Cho
- International Vaccine Institute, Seoul, Republic of Korea
| | - Amber Hsiao
- Technische Universität Berlin, Berlin, Germany
| | | | - T Anh Wartel
- International Vaccine Institute, Seoul, Republic of Korea
| | | | - Julia Lynch
- International Vaccine Institute, Seoul, Republic of Korea
| | - Jerome H Kim
- International Vaccine Institute, Seoul, Republic of Korea
| | | | | |
Collapse
|
15
|
Giannelli C, Raso MM, Palmieri E, De Felice A, Pippi F, Micoli F. Development of a Specific and Sensitive HPAEC-PAD Method for Quantification of Vi Polysaccharide Applicable to other Polysaccharides Containing Amino Uronic Acids. Anal Chem 2020; 92:6304-6311. [PMID: 32330386 DOI: 10.1021/acs.analchem.9b05107] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Typhoid fever is a major cause of morbidity and mortality in developing countries. Vaccines based on the Vi capsular polysaccharide are licensed or in development against typhoid fever. Vi content is a critical quality attribute for vaccines release, to monitor their stability and to ensure appropriate immune response. Vi polysaccharide is a homopolymer of α-1,4-N-acetylgalactosaminouronic acid, O-acetylated at the C-3 position, resistant to the commonly used acid hydrolysis for sugar chain depolymerization before monomer quantification. We previously developed a quantification method based on strong alkaline hydrolysis followed by High Performance Anion Exchange Chromatography-Pulsed Amperometric Detection analysis, but with low sensitivity and use for quantification of an unknown product coming from polysaccharide depolymerization. Here we describe the development of a method for Vi polysaccharide quantification based on acid hydrolysis with concomitant use of trifluoroacetic and hydrochloric acids. A Design of Experiment approach was used for the identification of the optimal hydrolysis conditions. The method is 100-fold more sensitive than the previous one, and specifically, resulting in the formation of a known product, confirmed to be the Vi monomer both de-O- and de-N-acetylated by mono- and bidimensional Nuclear Magnetic Resonance spectroscopy and mass spectrometry. Accuracy and precision were determined, and chromatographic conditions were improved to result in reduced time of analysis. This method will facilitate characterization of Vi-based vaccines. Furthermore, a similar approach has the potential to be extended to other polysaccharides containing 2-amino uronic acids, as already verified here for Shigella sonnei O-antigen, Streptococcus pneumoniae serotype 12F, and Staphylococcus aureus types 5 and 8 capsular polysaccharides.
Collapse
Affiliation(s)
- Carlo Giannelli
- GSK Vaccines Institute for Global Health, Via Fiorentina 1, 53100 Siena, Italy
| | | | - Elena Palmieri
- GSK Vaccines Institute for Global Health, Via Fiorentina 1, 53100 Siena, Italy
| | - Antonia De Felice
- GSK Vaccines Institute for Global Health, Via Fiorentina 1, 53100 Siena, Italy
| | - Federico Pippi
- GSK Vaccines Institute for Global Health, Via Fiorentina 1, 53100 Siena, Italy
| | - Francesca Micoli
- GSK Vaccines Institute for Global Health, Via Fiorentina 1, 53100 Siena, Italy
| |
Collapse
|
16
|
Sahastrabuddhe S, Saluja T. Overview of the Typhoid Conjugate Vaccine Pipeline: Current Status and Future Plans. Clin Infect Dis 2020; 68:S22-S26. [PMID: 30767002 PMCID: PMC6376107 DOI: 10.1093/cid/ciy884] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Typhoid fever remains a common and serious disease in populations that live in low- and middle-income countries. Treatment usually consists of antibiotics, but problems with drug-resistant strains have been increasing in endemic countries, making treatment prolonged and costly. Improved sanitation and food hygiene have been effective in controlling the disease in the industrialized world, but these steps are associated with socioeconomic progress that has been slow in most of the affected areas. Therefore, vaccination is an effective way to prevent the disease for the short to medium term. Oral typhoid vaccine and Vi polysaccharide typhoid vaccine (Vi polysaccharide) have been available for many years, yet a large population, in particular infants and children aged <2 years, remains at higher risk. Recently, with the availability of Vi polysaccharide–based conjugate vaccines and funding to support vaccination from the Gavi alliance, there is great momentum for typhoid prevention efforts. Supply of the vaccine will be critical, and there are multiple efforts to make new typhoid vaccines accessible and available to populations that desperately need them.
Collapse
Affiliation(s)
| | - Tarun Saluja
- International Vaccine Institute, Seoul, Republic of Korea
| |
Collapse
|
17
|
A phase 1 randomized safety, reactogenicity, and immunogenicity study of Typhax: A novel protein capsular matrix vaccine candidate for the prevention of typhoid fever. PLoS Negl Trop Dis 2020; 14:e0007912. [PMID: 31905228 PMCID: PMC6964911 DOI: 10.1371/journal.pntd.0007912] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 01/16/2020] [Accepted: 11/08/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Typhoid fever remains a significant cause of morbidity and mortality in developing countries especially in children ≤5 years old. Although the widely available unconjugated Vi polysaccharide vaccines are efficacious, they confer limited, short-term protection and are not approved for young children or infants. Vi conjugate vaccines, however, are now licensed in several typhoid endemic countries for use in children >6 months of age. As an alternative to conjugate vaccines, Matrivax has applied its novel 'virtual conjugation' Protein Capsular Matrix Vaccine (PCMV) technology to manufacture Typhax, which is composed of Vi polysaccharide entrapped in a cross-linked CRM197 matrix. METHODOLOGY A randomized, double-blinded, dose escalating Phase 1 study was performed to compare the safety and immunogenicity of three dose levels of aluminum phosphate adjuvanted Typhax (0.5, 2.5, or 10 μg of Vi antigen) to the FDA licensed vaccine, Typhim Vi, and placebo. Groups of 15 healthy adult subjects aged 18 to 55 years were randomized and received Typhax, Typhim Vi, or placebo at a ratio of 9:3:3. Typhax and placebo were administered in a two-dose regimen (Days 0 and 28) while Typhim Vi was administered as a single-dose on Day 0 with a placebo administered on Day 28. All doses were administered as a 0.5 mL intramuscular (IM) injection in a blinded fashion. The anti-Vi IgG antibody response was determined preimmunization (Day 0) and on Days 14, 28, 42, and 180 by ELISA. Seroconversion was defined as a titer 4-fold or greater above baseline. PRINCIPAL FINDINGS All Typhax vaccine regimens were well tolerated and adverse events were low in number and primarily characterized as mild in intensity and similar in incidence across the treatment groups. Reactogenicity, primarily pain and tenderness at the injection site, was observed in both the Typhax and Typhim Vi treatment groups; a modest increase in incidence was observed with increasing Typhax doses. Following one dose of Typhax, seroconversion rates at day 28 were 12.5%, 77.8%, 66.7% at the 0.5, 2.5, and 10 μg dose levels, respectively, compared to 55.6% and 0% in the Typhim Vi and placebo groups, respectively. A second dose of Typhax on Day 28 did not elicit a significant increase in GMT or seroconversion at Day 42 or Day 180 at any dose level. CONCLUSIONS Collectively, the results from this randomized phase 1 clinical trial indicate that Typhax is safe, well tolerated, and immunogenic. After a single dose, Typhax at the 2.5 and 10 μg dose levels elicited comparable anti-Vi IgG titers and seroconversion rates as a single dose of Typhim Vi (25 μg dose). A second dose of Typhax at Day 28 did not elicit a booster response. TRIAL REGISTRATION ClinicalTrials.gov NCT03926455.
Collapse
|
18
|
Meiring JE, Giubilini A, Savulescu J, Pitzer VE, Pollard AJ. Generating the Evidence for Typhoid Vaccine Introduction: Considerations for Global Disease Burden Estimates and Vaccine Testing Through Human Challenge. Clin Infect Dis 2019; 69:S402-S407. [PMID: 31612941 PMCID: PMC6792111 DOI: 10.1093/cid/ciz630] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Typhoid fever has had a major impact on human populations, with the causative pathogen Salmonella enterica serovar Typhi implicated in many outbreaks through history. The current burden of disease is estimated at 11-18 million infections annually, with the majority of infections located in Africa and South Asia. Data that have been used to estimate burden are limited to a small number of blood-culture surveillance studies, largely from densely populated urban centers. Extrapolating these data to estimate disease burden within and across countries highlights the lack of precision in global figures. A number of approaches have been developed, characterizing different geographical areas by water-based risk factors for typhoid infection or broader measures of health and development to more accurately extrapolate incidence. Recognition of the substantial disease burden is essential for policy-makers considering vaccine introduction. Typhoid vaccines have been in development for >100 years. The Vi polysaccharide (ViPS) and Ty21a vaccines have had a World Health Organization (WHO) recommendation for programmatic use in countries with high burden for 10 years, with 1 ViPS vaccine also having WHO prequalification. Despite this, uptake and introduction of these vaccines has been minimal. The development of a controlled human infection model (CHIM) enabled the accelerated testing of the newly WHO-prequalified ViPS-tetanus toxoid protein conjugate vaccine, providing efficacy estimates for the vaccine, prior to larger field trials. There is an urgency to the global control of enteric fever due to the escalating problem of antimicrobial resistance. With more accurate burden of disease estimates and a vaccine showing efficacy in CHIM, that control is now a possibility.
Collapse
Affiliation(s)
- James E Meiring
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, United Kingdom
- National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, United Kingdom
| | - Alberto Giubilini
- Oxford Uehiro Centre for Practical Ethics, University of Oxford, United Kingdom
| | - Julian Savulescu
- Oxford Uehiro Centre for Practical Ethics, University of Oxford, United Kingdom
| | - Virginia E Pitzer
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, Connecticut
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, United Kingdom
- National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, United Kingdom
| |
Collapse
|
19
|
Capeding MR, Alberto E, Sil A, Saluja T, Teshome S, Kim DR, Park JY, Yang JS, Chinaworapong S, Park J, Jo SK, Chon Y, Yang SY, Ham DS, Ryu JH, Lynch J, Kim JH, Kim H, Excler JL, Wartel TA, Sahastrabuddhe S. Immunogenicity, safety and reactogenicity of a Phase II trial of Vi-DT typhoid conjugate vaccine in healthy Filipino infants and toddlers: A preliminary report. Vaccine 2019; 38:4476-4483. [PMID: 31585725 PMCID: PMC7273193 DOI: 10.1016/j.vaccine.2019.09.074] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/20/2019] [Indexed: 11/28/2022]
Abstract
BACKGROUND Typhoid fever remains an important public health problem in developing countries and is endemic in many parts of Asia and Africa where the incidence of disease typically peaks in school-aged children. Age restrictions and other limitations of existing oral live-attenuated typhoid and parenteral Vi polysaccharide vaccines have triggered the development of Vi conjugate vaccines with improved immunological properties, use in younger age range, and longer durability of protection. We present the safety, reactogenicity, and immunogenicity data from a Phase II study after a single dose of Vi polysaccharide conjugated to diphtheria toxoid (Vi-DT) conducted in 6-23-month old Filipino children. METHODS This is a randomized, observer-blinded Phase II study to assess the immunogenicity, safety and reactogenicity of Vi-DT compared to placebo, conducted in Muntinlupa City, The Philippines. Participants aged 6-23 months were enrolled and randomized to Vi-DT (25 µg) or placebo (0.9% sodium chloride) and evaluated for immunogenicity and overall safety 28 days post vaccination. RESULTS A total of 285 participants were enrolled and age-stratified: 6 to < 9 months, 9-12 months, and 13-23 months. Seventy-six (76) participants received Vi-DT and 19 received placebo per each strata. All participants seroconverted after a single dose of Vi-DT versus 7% of placebo recipients. Anti-Vi IgG GMT was 444.38 [95% CI (400.28; 493.34)] after a single dose of Vi-DT; there was no change in GMT after placebo administration, 0.41 [95% CI (0.33; 0.51), p < 0.0001]. A similar pattern of immunogenicity was reported across all age strata. The vaccine reported to be safe and well tolerated. CONCLUSIONS Vi-DT vaccine was immunogenic, safe, and well tolerated in children aged 6-23 months. ClinicalTrials.gov registration number: NCT03527355.
Collapse
Affiliation(s)
| | - Edison Alberto
- Research Institute for Tropical Medicine, Manila, Philippines
| | - Arijit Sil
- International Vaccine Institute, Seoul, Republic of Korea
| | - Tarun Saluja
- International Vaccine Institute, Seoul, Republic of Korea.
| | - Samuel Teshome
- International Vaccine Institute, Seoul, Republic of Korea
| | - Deok Ryun Kim
- International Vaccine Institute, Seoul, Republic of Korea
| | - Ju Yeon Park
- International Vaccine Institute, Seoul, Republic of Korea
| | - Jae Seung Yang
- International Vaccine Institute, Seoul, Republic of Korea
| | | | - Jiwook Park
- International Vaccine Institute, Seoul, Republic of Korea
| | - Sue-Kyoung Jo
- International Vaccine Institute, Seoul, Republic of Korea
| | - Yun Chon
- International Vaccine Institute, Seoul, Republic of Korea
| | | | | | | | - Julia Lynch
- International Vaccine Institute, Seoul, Republic of Korea
| | - Jerome H Kim
- International Vaccine Institute, Seoul, Republic of Korea
| | - Hun Kim
- SK Bioscience, Seoul, Republic of Korea
| | | | - T Anh Wartel
- International Vaccine Institute, Seoul, Republic of Korea
| | | |
Collapse
|
20
|
Das S, Mohakud NK, Suar M, Sahu BR. Vaccine development for enteric bacterial pathogens: Where do we stand? Pathog Dis 2019; 76:5040763. [PMID: 30052916 DOI: 10.1093/femspd/fty057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 06/19/2018] [Indexed: 01/06/2023] Open
Abstract
Gut infections triggered by pathogenic bacteria lead to most frequently occurring diarrhea in humans accounting for million deaths annually. Currently, only a few licensed vaccines are available against these pathogens for mostly travelers moving to diarrheal endemic areas. Besides commercialized vaccines, there are many formulations that are either under clinical or pre-clinical stages of development and despite several efforts to improve safety, immunogenicity and efficacy, none of them can confer long-term protective immunity, for which repeated booster doses are always recommended. Further in many countries, financial, social and political constraints have jeopardized vaccine development program against these pathogens that enforce us to gather knowledge on safety, tolerability, immunogenicity and protective efficacy regarding the same. In this review, we analyze safety and efficacy issues of vaccines against five major gut bacteria causing enteric infections. The article also simultaneously describes several barriers for vaccine development and further discusses possible strategies to enhance immunogenicity and efficacy.
Collapse
Affiliation(s)
- Susmita Das
- Infection Biology Lab, KIIT School of Biotechnology, Campus XI, Bhubaneswar 751024, India
| | - Nirmal K Mohakud
- Department of Pediatrics, Kalinga Institute of Medical Sciences, Patia, Bhubaneswar 751024, India
| | - Mrutyunjay Suar
- Infection Biology Lab, KIIT School of Biotechnology, Campus XI, Bhubaneswar 751024, India
| | - Bikash R Sahu
- Infection Biology Lab, KIIT School of Biotechnology, Campus XI, Bhubaneswar 751024, India
| |
Collapse
|
21
|
Capeding MR, Teshome S, Saluja T, Syed KA, Kim DR, Park JY, Yang JS, Kim YH, Park J, Jo SK, Chon Y, Kothari S, Yang SY, Ham DS, Ryu JH, Hwang HS, Mun JH, Lynch JA, Kim JH, Kim H, Excler JL, Sahastrabuddhe S. Safety and immunogenicity of a Vi-DT typhoid conjugate vaccine: Phase I trial in Healthy Filipino adults and children. Vaccine 2018; 36:3794-3801. [PMID: 29776750 PMCID: PMC6005168 DOI: 10.1016/j.vaccine.2018.05.038] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/03/2018] [Accepted: 05/07/2018] [Indexed: 11/16/2022]
Abstract
BACKGROUND Typhoid fever remains a major public health problem in low- and middle-income countries where children aged 2-14 years bear the greatest burden. Vi polysaccharide is poorly immunogenic in children <2 years of age, and protection in adults is modest. The limitations of Vi polysaccharide vaccines can be overcome by conjugation of the Vi to a carrier protein. A typhoid conjugate vaccine composed of Vi polysaccharide conjugated to diphtheria toxoid (Vi-DT) has been developed. The Phase I study results are presented here. METHODS This was a randomized, observer-blinded Phase I study to assess the safety and immunogenicity of Vi-DT compared to Vi polysaccharide vaccine, conducted in Manila, Philippines. Participants enrolled in an age de-escalation manner (18-45, 6-17 and 2-5 years) were randomized between Test (Vi-DT, 25 µg) administered at 0 and 4 weeks and Comparator (Vi polysaccharide, Typhim Vi® and Vaxigrip®, Sanofi Pasteur) vaccines. RESULTS A total of 144 participants were enrolled (48 by age strata, 24 in Test and Comparator groups each). No serious adverse event was reported in either group. Solicited and unsolicited adverse events were mild or moderate in both groups with the exception of a 4-year old girl in Test group with grade 3 fever which resolved without sequelae. All participants in Test group seroconverted after first and second doses of Vi-DT while the proportions in the Comparator group were 97.1% and 97.2%, after first dose of Typhim Vi® and second dose of Vaxigrip®, respectively. Vi-DT showed 4-fold higher Geometric Mean Titers (GMT) compared to Typhim Vi® (adjusted for age strata, p < 0.001). No further increase of GMT was detected after the second dose of Vi-DT. Anti-DT IgG seroresponse rates were 81.2% and 84.5% post first and second Vi-DT doses, respectively. CONCLUSIONS Vi-DT vaccine was safe, well-tolerated and immunogenic in participants aged 2-45 years. ClinicalTrials.gov registration number: NCT02645032.
Collapse
Affiliation(s)
| | - Samuel Teshome
- International Vaccine Institute, Seoul, Republic of Korea
| | - Tarun Saluja
- International Vaccine Institute, Seoul, Republic of Korea.
| | | | - Deok Ryun Kim
- International Vaccine Institute, Seoul, Republic of Korea
| | - Ju Yeon Park
- International Vaccine Institute, Seoul, Republic of Korea
| | - Jae Seung Yang
- International Vaccine Institute, Seoul, Republic of Korea
| | - Yang Hee Kim
- International Vaccine Institute, Seoul, Republic of Korea
| | - Jiwook Park
- International Vaccine Institute, Seoul, Republic of Korea
| | - Sue-Kyoung Jo
- International Vaccine Institute, Seoul, Republic of Korea
| | - Yun Chon
- International Vaccine Institute, Seoul, Republic of Korea
| | - Sudeep Kothari
- International Vaccine Institute, Seoul, Republic of Korea
| | | | | | | | | | | | - Julia A Lynch
- International Vaccine Institute, Seoul, Republic of Korea
| | - Jerome H Kim
- International Vaccine Institute, Seoul, Republic of Korea
| | - Hun Kim
- SK Chemicals, Seoul, Republic of Korea
| | | | | |
Collapse
|
22
|
Abstract
BACKGROUND Typhoid fever and paratyphoid fever continue to be important causes of illness and death, particularly among children and adolescents in south-central and southeast Asia. Two typhoid vaccines are widely available, Ty21a (oral) and Vi polysaccharide (parenteral). Newer typhoid conjugate vaccines are at varying stages of development and use. The World Health Organization has recently recommended a Vi tetanus toxoid (Vi-TT) conjugate vaccine, Typbar-TCV, as the preferred vaccine for all ages. OBJECTIVES To assess the effects of vaccines for preventing typhoid fever. SEARCH METHODS In February 2018, we searched the Cochrane Infectious Diseases Group Specialized Register, CENTRAL, MEDLINE, Embase, LILACS, and mRCT. We also searched the reference lists of all included trials. SELECTION CRITERIA Randomized and quasi-randomized controlled trials (RCTs) comparing typhoid fever vaccines with other typhoid fever vaccines or with an inactive agent (placebo or vaccine for a different disease) in adults and children. Human challenge studies were not eligible. DATA COLLECTION AND ANALYSIS Two review authors independently applied inclusion criteria and extracted data, and assessed the certainty of the evidence using the GRADE approach. We computed vaccine efficacy per year of follow-up and cumulative three-year efficacy, stratifying for vaccine type and dose. The outcome addressed was typhoid fever, defined as isolation of Salmonella enterica serovar Typhi in blood. We calculated risk ratios (RRs) and efficacy (1 - RR as a percentage) with 95% confidence intervals (CIs). MAIN RESULTS In total, 18 RCTs contributed to the quantitative analysis in this review: 13 evaluated efficacy (Ty21a: 5 trials; Vi polysaccharide: 6 trials; Vi-rEPA: 1 trial; Vi-TT: 1 trial), and 9 reported on adverse events. All trials but one took place in typhoid-endemic countries. There was no information on vaccination in adults aged over 55 years of age, pregnant women, or travellers. Only one trial included data on children under two years of age.Ty21a vaccine (oral vaccine, three doses)A three-dose schedule of Ty21a vaccine probably prevents around half of typhoid cases during the first three years after vaccination (cumulative efficacy 2.5 to 3 years: 50%, 95% CI 35% to 61%, 4 trials, 235,239 participants, moderate-certainty evidence). These data include patients aged 3 to 44 years.Compared with placebo, this vaccine probably does not cause more vomiting, diarrhoea, nausea or abdominal pain (2 trials, 2066 participants; moderate-certainty evidence), headache, or rash (1 trial, 1190 participants; moderate-certainty evidence); however, fever (2 trials, 2066 participants; moderate-certainty evidence) is probably more common following vaccination.Vi polysaccharide vaccine (injection, one dose)A single dose of Vi polysaccharide vaccine prevents around two-thirds of typhoid cases in the first year after vaccination (year 1: 69%, 95% CI 63% to 74%; 3 trials, 99,979 participants; high-certainty evidence). In year 2, trial results were more variable, with the vaccine probably preventing between 45% and 69% of typhoid cases (year 2: 59%, 95% CI 45% to 69%; 4 trials, 194,969 participants; moderate-certainty evidence). These data included participants aged 2 to 55 years of age.The three-year cumulative efficacy of the vaccine may be around 55% (95% CI 30% to 70%; 11,384 participants, 1 trial; low-certainty evidence). These data came from a single trial conducted in South Africa in the 1980s in participants aged 5 to 15 years.Compared with placebo, this vaccine probably did not increase the incidence of fever (3 trials, 132,261 participants; moderate-certainty evidence) or erythema (3 trials, 132,261 participants; low-certainty evidence); however, swelling (3 trials, 1767 participants; moderate-certainty evidence) and pain at the injection site (1 trial, 667 participants; moderate-certainty evidence) were more common in the vaccine group.Vi-rEPA vaccine (two doses)Administration of two doses of the Vi-rEPA vaccine probably prevents between 50% and 96% of typhoid cases during the first two years after vaccination (year 1: 94%, 95% CI 75% to 99%; year 2: 87%, 95% CI 56% to 96%, 1 trial, 12,008 participants; moderate-certainty evidence). These data came from a single trial with children two to five years of age conducted in Vietnam.Compared with placebo, both the first and the second dose of this vaccine increased the risk of fever (1 trial, 12,008 and 11,091 participants, low-certainty evidence) and the second dose increase the incidence of swelling at the injection site (one trial, 11,091 participants, moderate-certainty evidence).Vi-TT vaccine (two doses)We are uncertain of the efficacy of administration of two doses of Vi-TT (PedaTyph) in typhoid cases in children during the first year after vaccination (year 1: 94%, 95% CI -1% to 100%, 1 trial, 1625 participants; very low-certainty evidence). These data come from a single cluster-randomized trial in children aged six months to 12 years and conducted in India. For single dose Vi-TT (Typbar-TCV), we found no efficacy trials evaluating the vaccine with natural exposure.There were no reported serious adverse effects in RCTs of any of the vaccines studied. AUTHORS' CONCLUSIONS The licensed Ty21a and Vi polysaccharide vaccines are efficacious in adults and children older than two years in endemic countries. The Vi-rEPA vaccine is just as efficacious, although data is only available for children. The new Vi-TT vaccine (PedaTyph) requires further evaluation to determine if it provides protection against typhoid fever. At the time of writing, there were only efficacy data from a human challenge setting in adults on the Vi-TT vaccine (Tybar), which clearly justify the ongoing field trials to evaluate vaccine efficacy.
Collapse
Affiliation(s)
- Rachael Milligan
- Liverpool School of Tropical MedicineCochrane Infectious Diseases GroupPembroke PlaceLiverpoolUKL3 5QA
| | - Mical Paul
- Rambam Health Care CampusDivision of Infectious DiseasesHa‐aliya 8 StHaifaIsrael33705
| | - Marty Richardson
- Liverpool School of Tropical MedicineCochrane Infectious Diseases GroupPembroke PlaceLiverpoolUKL3 5QA
| | - Ami Neuberger
- Rambam Health Care Campus and The Ruth and Bruce Rappaport Faculty of Medicine, Technion – Israel Institute of TechnologyDivision of Infectious DiseasesTel AvivIsrael
| |
Collapse
|
23
|
Arcuri M, Di Benedetto R, Cunningham AF, Saul A, MacLennan CA, Micoli F. The influence of conjugation variables on the design and immunogenicity of a glycoconjugate vaccine against Salmonella Typhi. PLoS One 2017; 12:e0189100. [PMID: 29287062 PMCID: PMC5747453 DOI: 10.1371/journal.pone.0189100] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 11/18/2017] [Indexed: 12/03/2022] Open
Abstract
In recent years there have been major efforts to develop glycoconjugate vaccines based on the Vi polysaccharide that will protect against Salmonella enterica Typhi infections, particularly typhoid fever, which remains a major public health concern in low-income countries. The design of glycoconjugate vaccines influences the immune responses they elicit. Here we systematically test the response in mice to Vi glycoconjugates that differ in Vi chain length (full-length and fragmented), carrier protein, conjugation chemistry, saccharide to protein ratio and size. We show that the length of Vi chains, but not the ultimate size of the conjugate, has an impact on the anti-Vi IgG immune response induced. Full-length Vi conjugates, independent of the carrier protein, induce peak IgG responses rapidly after just one immunization, and secondary immunization does not enhance the magnitude of these responses. Fragmented Vi linked to CRM197 and diphtheria toxoid, but not to tetanus toxoid, gives lower anti-Vi antibody responses after the first immunization than full-length Vi conjugates, but antibody titres are similar to those induced by full-length Vi conjugates following a second dose. The chemistry to conjugate Vi to the carrier protein, the linker used, and the saccharide to protein ratio do not significantly alter the response. We conclude that Vi length and carrier protein are the variables that influence the anti-Vi IgG response to immunization the most, while other parameters are of lesser importance.
Collapse
Affiliation(s)
- M. Arcuri
- GSK Vaccines Institute for Global Health (GVGH), Siena, Italy
- University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - R. Di Benedetto
- GSK Vaccines Institute for Global Health (GVGH), Siena, Italy
| | | | - A. Saul
- GSK Vaccines Institute for Global Health (GVGH), Siena, Italy
| | - C. A. MacLennan
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - F. Micoli
- GSK Vaccines Institute for Global Health (GVGH), Siena, Italy
| |
Collapse
|
24
|
Use of a novel antigen expressing system to study the Salmonella enterica serovar Typhi protein recognition by T cells. PLoS Negl Trop Dis 2017; 11:e0005912. [PMID: 28873442 PMCID: PMC5600385 DOI: 10.1371/journal.pntd.0005912] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/15/2017] [Accepted: 08/28/2017] [Indexed: 11/19/2022] Open
Abstract
Salmonella enterica serovar Typhi (S. Typhi), the causative agent of the typhoid fever, is a pathogen of great public health importance. Typhoid vaccines have the potential to be cost-effective measures towards combating this disease, yet the antigens triggering host protective immune responses are largely unknown. Given the key role of cellular-mediated immunity in S. Typhi protection, it is crucial to identify S. Typhi proteins involved in T-cell responses. Here, cells from individuals immunized with Ty21a typhoid vaccine were collected before and after immunization and used as effectors. We also used an innovative antigen expressing system based on the infection of B-cells with recombinant Escherichia coli (E. coli) expressing one of four S. Typhi gene products (i.e., SifA, OmpC, FliC, GroEL) as targets. Using flow cytometry, we found that the pattern of response to specific S. Typhi proteins was variable. Some individuals responded to all four proteins while others responded to only one or two proteins. We next evaluated whether T-cells responding to recombinant E. coli also possess the ability to respond to purified proteins. We observed that CD4+ cell responses, but not CD8+ cell responses, to recombinant E. coli were significantly associated with the responses to purified proteins. Thus, our results demonstrate the feasibility of using an E. coli expressing system to uncover the antigen specificity of T-cells and highlight its applicability to vaccine studies. These results also emphasize the importance of selecting the stimuli appropriately when evaluating CD4+ and CD8+ cell responses. Salmonella enterica serovar Typhi (S. Typhi) is the causative agent of the life-threatening typhoid fever that affects 11.9–20.6 million individuals annually in low-income and middle-income countries. The T-cells, CD4+ and CD8+ T cells, play a significant role in protection against S. Typhi infection. Yet, the antigens triggering host protective immune responses recognized by these cells are largely unknown. To address this shortcoming, in this study we used an E. coli expression system methodology for identifying immunogenic proteins of S. Typhi. We found that although the pattern of response to individual S. Typhi proteins was variable among the typhoid vaccinees, the E. coli expressing system uncovered the antigen specificity of T-cells, and highlight its applicability to vaccine studies.
Collapse
|
25
|
Giannelli C, Cappelletti E, Di Benedetto R, Pippi F, Arcuri M, Di Cioccio V, Martin L, Saul A, Micoli F. Determination of free polysaccharide in Vi glycoconjugate vaccine against typhoid fever. J Pharm Biomed Anal 2017; 139:143-147. [DOI: 10.1016/j.jpba.2017.02.042] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 02/13/2017] [Accepted: 02/20/2017] [Indexed: 10/20/2022]
|
26
|
Khatun F, Stephenson RJ, Toth I. An Overview of Structural Features of Antibacterial Glycoconjugate Vaccines That Influence Their Immunogenicity. Chemistry 2017; 23:4233-4254. [PMID: 28097690 DOI: 10.1002/chem.201603599] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Indexed: 12/13/2022]
Abstract
Bacterial cell-surface-derived or mimicked carbohydrate moieties that act as protective antigens are used in the development of antibacterial glycoconjugate vaccines. The carbohydrate antigen must have a minimum length or size to maintain the conformational structure of the antigenic epitope(s). The presence or absence of O-acetate, phosphate, glycerol phosphate and pyruvate ketal plays a vital role in defining the immunogenicity of the carbohydrate antigen. The nature of the carrier protein, spacer and conjugation pattern used to develop the glycoconjugate vaccine also defines its overall spatial orientation which in turn affects its avidity and selectivity of interaction with the desired target(s). In addition, the ratio of carbohydrate to protein in glycoconjugate vaccines also makes an important contribution in determining the optimum immunological response. This Review article presents the importance of these variables in the development of antibacterial glycoconjugate vaccines and their effects on immune efficacy.
Collapse
Affiliation(s)
- Farjana Khatun
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia
| | - Rachel J Stephenson
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia.,School of Pharmacy, Woolloongabba, The University of Queensland, QLD, Australia.,Institute for Molecular Bioscience, St. Lucia, The University of Queensland, QLD, Australia
| |
Collapse
|
27
|
Jang MS, Sahastrabuddhe S, Yun CH, Han SH, Yang JS. Serum bactericidal assay for the evaluation of typhoid vaccine using a semi-automated colony-counting method. Microb Pathog 2016; 97:19-26. [PMID: 27216239 PMCID: PMC4944902 DOI: 10.1016/j.micpath.2016.05.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 05/18/2016] [Accepted: 05/19/2016] [Indexed: 12/22/2022]
Abstract
Typhoid fever, mainly caused by Salmonella enterica serovar Typhi (S. Typhi), is a life-threatening disease, mostly in developing countries. Enzyme-linked immunosorbent assay (ELISA) is widely used to quantify antibodies against S. Typhi in serum but does not provide information about functional antibody titers. Although the serum bactericidal assay (SBA) using an agar plate is often used to measure functional antibody titers against various bacterial pathogens in clinical specimens, it has rarely been used for typhoid vaccines because it is time-consuming and labor-intensive. In the present study, we established an improved SBA against S. Typhi using a semi-automated colony-counting system with a square agar plate harboring 24 samples. The semi-automated SBA efficiently measured bactericidal titers of sera from individuals immunized with S. Typhi Vi polysaccharide vaccines. The assay specifically responded to S. Typhi Ty2 but not to other irrelevant enteric bacteria including Vibrio cholerae and Shigella flexneri. Baby rabbit complement was more appropriate source for the SBA against S. Typhi than complements from adult rabbit, guinea pig, and human. We also examined the correlation between SBA and ELISA for measuring antibody responses against S. Typhi using pre- and post-vaccination sera from 18 human volunteers. The SBA titer showed a good correlation with anti-Vi IgG quantity in the serum as determined by Spearman correlation coefficient of 0.737 (P < 0.001). Taken together, the semi-automated SBA might be efficient, accurate, sensitive, and specific enough to measure functional antibody titers against S. Typhi in sera from human subjects immunized with typhoid vaccines. Improved SBA against S. Typhi was developed using a colony counting system. The improved SBA was specific to S. Typhi but not to other gram-negative bacteria. There was a good correlation between SBA and anti-Vi IgG titers in vaccinee’s sera. This SBA would be useful for the clinical immuno-monitoring of typhoid vaccines.
Collapse
Affiliation(s)
- Mi Seon Jang
- Clinical Immunology, Sciences Unit, International Vaccine Institute, Seoul, Republic of Korea; Department of Oral Microbiology and Immunology, DRI, and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | | | - Cheol-Heui Yun
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, DRI, and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul, Republic of Korea.
| | - Jae Seung Yang
- Clinical Immunology, Sciences Unit, International Vaccine Institute, Seoul, Republic of Korea.
| |
Collapse
|
28
|
Leroux-Roels G, Maes C, Willekens J, De Boever F, de Rooij R, Martell L, Bedell L, Wittke F, Slobod K, Dull P. A randomized, observer-blind Phase Ib study to identify formulations and vaccine schedules of a trivalent Group B Streptococcus vaccine for use in non-pregnant and pregnant women. Vaccine 2016; 34:1786-91. [DOI: 10.1016/j.vaccine.2016.02.044] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/12/2016] [Accepted: 02/16/2016] [Indexed: 11/24/2022]
|
29
|
Czerkinsky C, Holmgren J. Vaccines against enteric infections for the developing world. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2015.0142. [PMID: 25964464 DOI: 10.1098/rstb.2015.0142] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Since the first licensure of the Sabin oral polio vaccine more than 50 years ago, only eight enteric vaccines have been licensed for four disease indications, and all are given orally. While mucosal vaccines offer programmatically attractive tools for facilitating vaccine deployment, their development remains hampered by several factors: -limited knowledge regarding the properties of the gut immune system during early life; -lack of mucosal adjuvants, limiting mucosal vaccine development to live-attenuated or killed whole virus and bacterial vaccines; -lack of correlates/surrogates of mucosal immune protection; and -limited knowledge of the factors contributing to oral vaccine underperformance in children from developing countries. There are now reasons to believe that the development of safe and effective mucosal adjuvants and of programmatically sound intervention strategies could enhance the efficacy of current and next-generation enteric vaccines, especially in lesser developed countries which are often co-endemic for enteric infections and malnutrition. These vaccines must be safe and affordable for the world's poorest, confer long-term protection and herd immunity, and must be able to contain epidemics.
Collapse
Affiliation(s)
- Cecil Czerkinsky
- CNRS, INSERM, Institut de Pharmacologie Moleculaire et Cellulaire, Université Nice Sophia Antipolis, 06103 Nice, France
| | - Jan Holmgren
- Department of Microbiology and Immunology, University of Gothenburg Vaccine Research Institute (GUVAX), Gothenburg 405 30, Sweden
| |
Collapse
|
30
|
Hatz CFR, Bally B, Rohrer S, Steffen R, Kramme S, Siegrist CA, Wacker M, Alaimo C, Fonck VG. Safety and immunogenicity of a candidate bioconjugate vaccine against Shigella dysenteriae type 1 administered to healthy adults: A single blind, partially randomized Phase I study. Vaccine 2015; 33:4594-601. [PMID: 26162850 DOI: 10.1016/j.vaccine.2015.06.102] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 06/01/2015] [Accepted: 06/26/2015] [Indexed: 11/18/2022]
Abstract
BACKGROUND Shigellae cause severe disease in endemic countries, especially in children. Several efficacy trials have been conducted with candidate vaccines against Shigellae, but the lack of protection, the safety concerns, or manufacturing challenges hindered successful market approval. Conjugated vaccines have been shown to be safe and effective for different pathogens (i.e., Neisseria meningitidis, Shigella pneumonia, Haemophilus influenzae). The bio-conjugation technology, exploited here for the Shigella dysenteriae candidate vaccine, offers a novel and potentially simpler way to develop and produce vaccines against one of the major causes of morbidity and mortality in developing countries. METHODS A novel S. dysenteriae bioconjugate vaccine (GVXN SD133) made of the polysaccharide component of the Shigella O1 lipopolysaccharide, conjugated to the exotoxin protein A of Pseudomonas aeruginosa (EPA), was evaluated for immunogenicity and safety in healthy adults in a single blind, partially randomized Phase I study. Forty subjects (10 in each dose group; 2 μg or 10 μg with or without aluminium adjuvant) received two injections 60 days apart and were followed-up for 150 days. RESULTS Both doses and formulations were well tolerated; the safety and reactogenicity profiles were consistent with that of other conjugated vaccines, adjuvanted or not, independent of the dose and the number of injections. The GVXN SD133 vaccine elicited statistically significant O1 specific humoral responses at all time points in all vaccination groups. Between-group comparisons did not show statistically significant differences in geometric mean titers of immunoglobulin G and A at any post-vaccination time point. CONCLUSIONS This study demonstrated that the GVXN SD133 vaccine has a satisfactory safety profile. It elicited a significant humoral response to Shigella O1 polysaccharides at all doses tested. The protein carrier also elicited functional antibodies, showing the technology's advantages in preserving both sugar and conjugated protein epitopes. This trial is registered at ClinicalTrials.gov (NCT01069471).
Collapse
Affiliation(s)
- Christoph F R Hatz
- Epidemiology, Biostatistics and Prevention Institute (formerly Social and Preventive Medicine (ISPM), University of Zurich, Hirschengraben 84, Zurich 8001, Switzerland; Swiss Tropical and Public Health Institute, 4002 Basel, Switzerland
| | - Bettina Bally
- Epidemiology, Biostatistics and Prevention Institute (formerly Social and Preventive Medicine (ISPM), University of Zurich, Hirschengraben 84, Zurich 8001, Switzerland
| | - Susanne Rohrer
- GlycoVaxyn AG, Grabenstrasse 3, Schlieren 8952, Switzerland
| | - Robert Steffen
- Epidemiology, Biostatistics and Prevention Institute (formerly Social and Preventive Medicine (ISPM), University of Zurich, Hirschengraben 84, Zurich 8001, Switzerland
| | - Stefanie Kramme
- Swiss Tropical and Public Health Institute, 4002 Basel, Switzerland
| | - Claire-Anne Siegrist
- Center for Vaccinology, University of Geneva, CMU, 1 Michel-Servet, Geneva 4, Geneva 1211, Switzerland
| | - Michael Wacker
- GlycoVaxyn AG, Grabenstrasse 3, Schlieren 8952, Switzerland
| | | | | |
Collapse
|
31
|
Abstract
Control of typhoid fever relies on clinical information, diagnosis, and an understanding for the epidemiology of the disease. Despite the breadth of work done so far, much is not known about the biology of this human-adapted bacterial pathogen and the complexity of the disease in endemic areas, especially those in Africa. The main barriers to control are vaccines that are not immunogenic in very young children and the development of multidrug resistance, which threatens efficacy of antimicrobial chemotherapy. Clinicians, microbiologists, and epidemiologists worldwide need to be familiar with shifting trends in enteric fever. This knowledge is crucial, both to control the disease and to manage cases. Additionally, salmonella serovars that cause human infection can change over time and location. In areas of Asia, multidrug-resistant Salmonella enterica serovar Typhi (S Typhi) has been the main cause of enteric fever, but now S Typhi is being displaced by infections with drug-resistant S enterica serovar Paratyphi A. New conjugate vaccines are imminent and new treatments have been promised, but the engagement of local medical and public health institutions in endemic areas is needed to allow surveillance and to implement control measures.
Collapse
Affiliation(s)
- John Wain
- Norwich Medical School, University of East Anglia, Norwich, UK.
| | - Rene S Hendriksen
- National Food Institute, Technical University of Denmark, WHO Collaborating Centre for Antimicrobial Resistance in Foodborne Pathogens and European Union Reference Laboratory for Antimicrobial Resistance, Kongens Lyngby, Denmark
| | - Matthew L Mikoleit
- National Enteric Reference Laboratory Team, Enteric Diseases Laboratory Branch, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Karen H Keddy
- Centre for Enteric Diseases, National Institute for Communicable Diseases, Division in the National Health Laboratory Service (NHLS), Johannesburg, South Africa; Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | |
Collapse
|
32
|
Dewé W, Durand C, Marion S, Oostvogels L, Devaster JM, Fourneau M. A multi-criteria decision making approach to identify a vaccine formulation. J Biopharm Stat 2015; 26:352-64. [PMID: 25616785 DOI: 10.1080/10543406.2015.1008517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
This article illustrates the use of a multi-criteria decision making approach, based on desirability functions, to identify an appropriate adjuvant composition for an influenza vaccine to be used in elderly. The proposed adjuvant system contained two main elements: monophosphoryl lipid and α-tocopherol with squalene in an oil/water emulsion. The objective was to elicit a stronger immune response while maintaining an acceptable reactogenicity and safety profile. The study design, the statistical models, the choice of the desirability functions, the computation of the overall desirability index, and the assessment of the robustness of the ranking are all detailed in this manuscript.
Collapse
Affiliation(s)
- Walthère Dewé
- a Biostatistics, GlaxoSmithKline Vaccines , Rixensart , Belgium
| | | | | | | | | | - Marc Fourneau
- a Biostatistics, GlaxoSmithKline Vaccines , Rixensart , Belgium
| |
Collapse
|
33
|
|
34
|
Sztein MB, Salerno-Goncalves R, McArthur MA. Complex adaptive immunity to enteric fevers in humans: lessons learned and the path forward. Front Immunol 2014; 5:516. [PMID: 25386175 PMCID: PMC4209864 DOI: 10.3389/fimmu.2014.00516] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 10/03/2014] [Indexed: 01/26/2023] Open
Abstract
Salmonella enterica serovar Typhi (S. Typhi), the causative agent of typhoid fever, and S. Paratyphi A and B, causative agents of paratyphoid fever, are major public health threats throughout the world. Although two licensed typhoid vaccines are currently available, they are only moderately protective and immunogenic necessitating the development of novel vaccines. A major obstacle in the development of improved typhoid, as well as paratyphoid vaccines is the lack of known immunological correlates of protection in humans. Considerable progress has been made in recent years in understanding the complex adaptive host responses against S. Typhi. Although the induction of S. Typhi-specific antibodies (including their functional properties) and memory B cells, as well as their cross-reactivity with S. Paratyphi A and S. Paratyphi B has been shown, the role of humoral immunity in protection remains undefined. Cell mediated immunity (CMI) is likely to play a dominant role in protection against enteric fever pathogens. Detailed measurements of CMI performed in volunteers immunized with attenuated strains of S. Typhi have shown, among others, the induction of lymphoproliferation, multifunctional type 1 cytokine production, and CD8(+) cytotoxic T-cell responses. In addition to systemic responses, the local microenvironment of the gut is likely to be of paramount importance in protection from these infections. In this review, we will critically assess current knowledge regarding the role of CMI and humoral immunity following natural S. Typhi and S. Paratyphi infections, experimental challenge, and immunization in humans. We will also address recent advances regarding cross-talk between the host's gut microbiota and immunization with attenuated S. Typhi, mechanisms of systemic immune responses, and the homing potential of S. Typhi-specific B- and T-cells to the gut and other tissues.
Collapse
Affiliation(s)
- Marcelo B Sztein
- Department of Pediatrics, Center for Vaccine Development (CVD), University of Maryland School of Medicine , Baltimore, MD , USA
| | - Rosangela Salerno-Goncalves
- Department of Pediatrics, Center for Vaccine Development (CVD), University of Maryland School of Medicine , Baltimore, MD , USA
| | - Monica A McArthur
- Department of Pediatrics, Center for Vaccine Development (CVD), University of Maryland School of Medicine , Baltimore, MD , USA
| |
Collapse
|
35
|
Coward C, Restif O, Dybowski R, Grant AJ, Maskell DJ, Mastroeni P. The effects of vaccination and immunity on bacterial infection dynamics in vivo. PLoS Pathog 2014; 10:e1004359. [PMID: 25233077 PMCID: PMC4169467 DOI: 10.1371/journal.ppat.1004359] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 07/25/2014] [Indexed: 01/31/2023] Open
Abstract
Salmonella enterica infections are a significant global health issue, and development of vaccines against these bacteria requires an improved understanding of how vaccination affects the growth and spread of the bacteria within the host. We have combined in vivo tracking of molecularly tagged bacterial subpopulations with mathematical modelling to gain a novel insight into how different classes of vaccines and branches of the immune response protect against secondary Salmonella enterica infections of the mouse. We have found that a live Salmonella vaccine significantly reduced bacteraemia during a secondary challenge and restrained inter-organ spread of the bacteria in the systemic organs. Further, fitting mechanistic models to the data indicated that live vaccine immunisation enhanced both the bacterial killing in the very early stages of the infection and bacteriostatic control over the first day post-challenge. T-cell immunity induced by this vaccine is not necessary for the enhanced bacteriostasis but is required for subsequent bactericidal clearance of Salmonella in the blood and tissues. Conversely, a non-living vaccine while able to enhance initial blood clearance and killing of virulent secondary challenge bacteria, was unable to alter the subsequent bacterial growth rate in the systemic organs, did not prevent the resurgence of extensive bacteraemia and failed to control the spread of the bacteria in the body. The bacterium Salmonella enterica causes gastroenteritis and the severe systemic diseases typhoid, paratyphoid fever and non-typhoidal septicaemia (NTS). Treatment of systemic disease with antibiotics is becoming increasingly difficult due to the acquisition of resistance. Licensed vaccines are available for the prevention of typhoid, but not paratyphoid fever or NTS. Vaccines can be either living (attenuated strains) or non-living (e.g. inactivated whole cells or surface polysaccharides) and these different classes potentially activate different components of the host immune system. Improvements in vaccine design require a better understanding of how different vaccine types differ in their ability to control a subsequent infection. We have improved a previously developed experimental system and mathematical model to investigate how these different vaccine types act. We show that the inactivated vaccine can only control bacterial numbers by a transient increase in bactericidal activity whereas the living vaccine is superior as it can induce an immune response that rapidly kills, then restrains the growth and spread of infecting bacteria.
Collapse
Affiliation(s)
- Chris Coward
- University of Cambridge, Department of Veterinary Medicine, Cambridge, United Kingdom
| | - Olivier Restif
- University of Cambridge, Department of Veterinary Medicine, Cambridge, United Kingdom
| | - Richard Dybowski
- University of Cambridge, Department of Veterinary Medicine, Cambridge, United Kingdom
| | - Andrew J Grant
- University of Cambridge, Department of Veterinary Medicine, Cambridge, United Kingdom
| | - Duncan J Maskell
- University of Cambridge, Department of Veterinary Medicine, Cambridge, United Kingdom
| | - Pietro Mastroeni
- University of Cambridge, Department of Veterinary Medicine, Cambridge, United Kingdom
| |
Collapse
|
36
|
MacLennan CA, Martin LB, Micoli F. Vaccines against invasive Salmonella disease: current status and future directions. Hum Vaccin Immunother 2014; 10:1478-93. [PMID: 24804797 PMCID: PMC4185946 DOI: 10.4161/hv.29054] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Though primarily enteric pathogens, Salmonellae are responsible for a considerable yet under-appreciated global burden of invasive disease. In South and South-East Asia, this manifests as enteric fever caused by serovars Typhi and Paratyphi A. In sub-Saharan Africa, a similar disease burden results from invasive nontyphoidal Salmonellae, principally serovars Typhimurium and Enteritidis. The existing Ty21a live-attenuated and Vi capsular polysaccharide vaccines target S. Typhi and are not effective in young children where the burden of invasive Salmonella disease is highest. After years of lack of investment in new Salmonella vaccines, recent times have seen increased interest in the area led by emerging-market manufacturers, global health vaccine institutes and academic partners. New glycoconjugate vaccines against S. Typhi are becoming available with similar vaccines against other invasive serovars in development. With other new vaccines under investigation, including live-attenuated, protein-based and GMMA vaccines, now is an exciting time for the Salmonella vaccine field.
Collapse
Affiliation(s)
- Calman A MacLennan
- Novartis Vaccines Institute for Global Health; Siena, Italy; Medical Research Council Centre for Immune Regulation and Clinical Immunology Service; Institute of Biomedical Research, School of Immunity and Infection; College of Medicine and Dental Sciences; University of Birmingham; Birmingham, UK
| | - Laura B Martin
- Novartis Vaccines Institute for Global Health; Siena, Italy
| | | |
Collapse
|
37
|
Sahastrabuddhe S, Carbis R, Wierzba TF, Ochiai RL. Increasing rates of Salmonella Paratyphi A and the current status of its vaccine development. Expert Rev Vaccines 2014; 12:1021-31. [PMID: 24053396 DOI: 10.1586/14760584.2013.825450] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Enteric fever caused by Salmonella enterica serovar Typhi and Salmonella enterica serovar Paratyphi is still a major disease burden mainly in developing countries. Previously, S. Typhi was believed to be the major cause of enteric fever. The real situation is now becoming clear with reports emerging from many Asian countries of S. Paratyphi, mostly S. Paratyphi A, causing a substantial number of cases of enteric fever. Although there have been advances in the use of the currently available typhoid vaccines and in the development of newer typhoid vaccines, paratyphoid vaccine development is lagging behind. Since the disease caused by S. Typhi and S. Paratyphi are clinically indistinguishable and are commonly termed 'enteric' fever, it will be necessary to have a vaccine available against both S. Typhi and S. Paratyphi A as a bivalent 'enteric fever vaccine'.
Collapse
Affiliation(s)
- Sushant Sahastrabuddhe
- International Vaccine Institute, San 4-8, Nakseongdae-dong, Gwanak-gu, Seoul, 151-919, Korea
| | | | | | | |
Collapse
|
38
|
|
39
|
Steele D, Riddle M, van de Verg L, Bourgeois L. Vaccines for enteric diseases: a meeting summary. Expert Rev Vaccines 2014; 11:407-9. [DOI: 10.1586/erv.12.9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
40
|
Abstract
BACKGROUND Typhoid fever and paratyphoid fever continue to be important causes of illness and death, particularly among children and adolescents in south-central and southeast Asia. Two typhoid vaccines are commercially available, Ty21a (oral) and Vi polysaccharide (parenteral), but neither is used routinely. Other vaccines, such as a new, modified, conjugated Vi vaccine called Vi-rEPA, are in development. OBJECTIVES To evaluate the efficacy and adverse effects of vaccines used to prevent typhoid fever. SEARCH METHODS In June 2013, we searched the Cochrane Infectious Diseases Group Specialized Register, CENTRAL, MEDLINE, EMBASE, LILACS, and mRCT. We also searched relevant conference proceedings up to 2013 and scanned the reference lists of all included trials. SELECTION CRITERIA Randomized and quasi-randomized controlled trials (RCTs) comparing typhoid fever vaccines with other typhoid fever vaccines or with an inactive agent (placebo or vaccine for a different disease). DATA COLLECTION AND ANALYSIS Two review authors independently applied inclusion criteria and extracted data. We computed vaccine efficacy per year of follow-up and cumulative three-year efficacy, stratifying for vaccine type and dose. The outcome addressed was typhoid fever, defined as isolation of Salmonella typhi in blood. We calculated risk ratios (RRs) and efficacy (1-RR as a percentage) with 95% confidence intervals (CIs). MAIN RESULTS In total, 18 RCTs were included in this review; 12 evaluated efficacy (Ty21a: five trials; Vi polysaccharide: six trials; Vi-rEPA: one trial), and 11 reported on adverse events. Ty21a vaccine (oral vaccine, three doses) A three-dose schedule of Ty21a vaccine prevents around one-third to one-half of typhoid cases in the first two years after vaccination (Year 1: 35%, 95% CI 8% to 54%; Year 2: 58%, 95% CI 40% to 71%; one trial, 20,543 participants; moderate quality evidence; data taken from a single trial conducted in Indonesia in the 1980s). No benefit was detected in the third year after vaccination. Four additional cluster-RCTs have been conducted, but the study authors did not adjust for clustering.Compared with placebo, this vaccine was not associated with more participants with vomiting, diarrhoea, nausea or abdominal pain (four trials, 2066 participants; moderate quality evidence) headache, or rash (two trials, 1190 participants; moderate quality evidence); however, fever (four trials, 2066 participants; moderate quality evidence) was more common in the vaccine group. Vi polysaccharide vaccine (injection, one dose) A single dose of Vi polysaccharide vaccine prevents around two-thirds of typhoid cases in the first year after vaccination (Year 1: 69%, 95% CI 63% to 74%; three trials, 99,979 participants; high quality evidence). In Year 2, the trial results were more variable, with the vaccine preventing between 45% and 69% of typhoid cases (Year 2: 59%, 95% CI 45% to 69%; four trials, 194,969 participants; moderate quality evidence). The three-year cumulative efficacy of the vaccine is around 55% (95% CI 30% to 70%; 11,384 participants, one trial; moderate quality evidence). These data are taken from a single trial in South Africa in the 1980s.Compared with placebo, this vaccine was not associated with more participants with fever (four trials, 133,038 participants; moderate quality evidence) or erythema (three trials, 132,261 participants; low quality evidence); however, swelling (three trials, 1767 participants; moderate quality evidence) and pain at the injection site (one trial, 667 participants; moderate quality evidence) were more common in the vaccine group. Vi-rEPA vaccine (two doses) Administration of two doses of the Vi-rEPA vaccine prevents between 50% and 96% of typhoid cases during the first two years after vaccination (Year 1: 94%, 95% CI 75% to 99%; Year 2: 87%, 95% CI 56% to 96%; one trial, 12,008 participants; moderate quality evidence). These data are taken from a single trial with children 2 to 5 years of age conducted in Vietnam.Compared with placebo, the first and second doses of this vaccine were not associated with increased risk of adverse events. The first dose of this vaccine was not associated with fever (2 studies, 12,209 participants; low quality evidence), erythema (two trials, 12,209 participants; moderate quality evidence) or swelling at the injection site (two trials, 12,209 participants; moderate quality evidence). The second dose of this vaccine was not associated with fever (two trials, 11,286 participants; low quality evidence), erythema (two trials, 11,286 participants; moderate quality evidence) and swelling at the injection site (two trials, 11,286 participants; moderate quality evidence). AUTHORS' CONCLUSIONS The licensed Ty21a and Vi polysaccharide vaccines are efficacious. The new and unlicensed Vi-rEPA vaccine is as efficacious and may confer longer immunity.
Collapse
Affiliation(s)
- Elspeth Anwar
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | | | | | | | | | | |
Collapse
|
41
|
Waddington CS, Darton TC, Pollard AJ. The challenge of enteric fever. J Infect 2014; 68 Suppl 1:S38-50. [DOI: 10.1016/j.jinf.2013.09.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2013] [Indexed: 11/27/2022]
|
42
|
Bhutta ZA, Capeding MR, Bavdekar A, Marchetti E, Ariff S, Soofi SB, Anemona A, Habib MA, Alberto E, Juvekar S, Khan RMQ, Marhaba R, Ali N, Malubay N, Kawade A, Saul A, Martin LB, Podda A. Immunogenicity and safety of the Vi-CRM197 conjugate vaccine against typhoid fever in adults, children, and infants in south and southeast Asia: results from two randomised, observer-blind, age de-escalation, phase 2 trials. THE LANCET. INFECTIOUS DISEASES 2013; 14:119-29. [PMID: 24290843 DOI: 10.1016/s1473-3099(13)70241-x] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Typhoid vaccination is a public health priority in developing countries where young children are greatly affected by typhoid fever. Because present vaccines are not recommended for children younger than 2 years, the Novartis Vaccines Institute for Global Health developed a conjugate vaccine (Vi-CRM197) for infant immunisation. We aimed to assess the immunogenicity and safety of Vi-CRM197 in participants of various ages in endemic countries in south and southeast Asia. METHODS We did two randomised, observer-blind, age de-escalation, phase 2 trials at two sites in Pakistan and India (study A), and at one site in the Philippines (study B), between March 2, 2011, and Aug 9, 2012. Adults aged 18-45 years, children aged 24-59 months, older infants aged 9-12 months, and infants aged 6-8 weeks were randomly assigned (1:1) with a computer-generated randomisation list (block size of four) to receive either 5 μg Vi-CRM197 or 25 μg Vi-polysaccharide vaccine (or 13-valent pneumococcal conjugate vaccine in children younger than 2 years). Both infant populations received Vi-CRM197 concomitantly with vaccines of the Expanded Programme on Immunization (EPI), according to WHO schedule. With the exception of designated study site personnel responsible for vaccine preparation, study investigators, those assessing outcomes, and data analysts were masked to treatment allocation. We specified no a-priori null hypothesis for the immunogenicity or safety objectives and all analyses were descriptive. Analyses were by modified intention-to-treat. These studies are registered with ClinicalTrials.gov, numbers NCT01229176 and NCT01437267. FINDINGS 320 participants were enrolled and vaccinated in the two trials: 200 in study A (all age groups) and 120 in study B (children and infants only), of whom 317 (99%) were included in the modified intention-to-treat analysis. One dose of Vi-CRM197 significantly increased concentrations of anti-Vi antibody in adults (from 113 U/mL [95% CI 67-190] to 208 U/mL [117-369]), children (201 U/mL [138-294] to 368 U/mL [234-580]), and older infants (179 U/mL [129-250] to 249 U/mL [130-477]). However, in children and older infants, a second dose of conjugate vaccine had no incremental effect on antibody titres and, at all ages, concentrations of antibodies increased substantially 6 months after vaccination (from 55 U/mL [33-94] to 63 U/mL [35-114] in adults, from 23 U/mL [15-34] to 51 U/mL [34-76] in children, and from 21 U/mL [14-31] to 22 U/mL [14-33] in older infants). Immune response in infants aged 6-8 weeks was lower than that in older participants and, 6 months after third vaccination, antibody concentrations were significantly higher than pre-vaccination concentrations in Filipino (21 U/mL [16-28] vs 2.88 U/mL [1.95-4.25]), but not Pakistani (3.76 U/mL [2.77-5.08] vs 2.77 U/mL [2.1-3.66]), infants. Vi-CRM197 was safe and well tolerated and did not induce any significant interference with EPI vaccines. No deaths or vaccine-related serious adverse events were reported throughout the studies. INTERPRETATION Vi-CRM197 is safe and immunogenic in endemic populations of all ages. Given at 9 months of age, concomitantly with measles vaccine, Vi-CRM197 shows a promise for potential inclusion in EPI schedules of countries endemic for typhoid. An apparent absence of booster response and a reduction in antibody titres 6 months after immunisation should be further investigated, but data show that an immunogenic typhoid vaccine can be safely delivered to infants during EPI visits recommended by WHO. FUNDING Sclavo Vaccines Association and Regione Toscana.
Collapse
Affiliation(s)
- Zulfiqar A Bhutta
- Department of Pediatrics and Child Health, Division of Women and Child Health, Aga Khan University, Karachi, Pakistan
| | | | - Ashish Bavdekar
- King Edward Memorial Hospital Research Centre, Rasta Peth, Pune, India
| | | | - Shabina Ariff
- Novartis Vaccines Institute for Global Health, Siena, Italy
| | - Sajid B Soofi
- Department of Pediatrics and Child Health, Division of Women and Child Health, Aga Khan University, Karachi, Pakistan
| | | | - Muhammad A Habib
- Department of Pediatrics and Child Health, Division of Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Edison Alberto
- Research Institute for Tropical Medicine, Manila, Philippines
| | - Sanjay Juvekar
- King Edward Memorial Hospital Research Centre, Rasta Peth, Pune, India
| | | | - Rachid Marhaba
- Novartis Vaccines and Diagnostics Human Serology Laboratories, Marburg, Germany
| | - Noshad Ali
- Department of Pediatrics and Child Health, Division of Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Nelia Malubay
- Research Institute for Tropical Medicine, Manila, Philippines
| | - Anand Kawade
- King Edward Memorial Hospital Research Centre, Rasta Peth, Pune, India
| | - Allan Saul
- Novartis Vaccines Institute for Global Health, Siena, Italy
| | - Laura B Martin
- Novartis Vaccines Institute for Global Health, Siena, Italy
| | - Audino Podda
- Novartis Vaccines Institute for Global Health, Siena, Italy.
| |
Collapse
|
43
|
Saul A, Smith T, Maire N. Stochastic simulation of endemic Salmonella enterica serovar Typhi: the importance of long lasting immunity and the carrier state. PLoS One 2013; 8:e74097. [PMID: 24040177 PMCID: PMC3769365 DOI: 10.1371/journal.pone.0074097] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 07/28/2013] [Indexed: 11/29/2022] Open
Abstract
Background Typhoid fever caused by Salmonella enterica serovar Typhi (S. Typhi) remains a serious burden of disease, especially in developing countries of Asia and Africa. It is estimated that it causes 200,000 deaths per year, mainly in children. S. Typhi is an obligate pathogen of humans and although it has a relatively complex life cycle with a long lived carrier state, the absence of non-human hosts suggests that well targeted control methods should have a major impact on disease. Newer control methods including new generations of vaccines offer hope but their implementation would benefit from quantitative models to guide the most cost effective strategies. This paper presents a quantitative model of Typhoid disease, immunity and transmission as a first step in that process. Methodology/Principal Findings A stochastic agent-based model has been developed that incorporates known features of the biology of typhoid including probability of infection, the consequences of infection, treatment options, acquisition and loss of immunity as a result of infection and vaccination, the development of the carrier state and the impact of environmental or behavioral factors on transmission. The model has been parameterized with values derived where possible from the literature and where this was not possible, feasible parameters space has been determined by sensitivity analyses, fitting the simulations to age distribution of field data. The model is able to adequately predict the age distribution of typhoid in two settings. Conclusions/Significance The modeling highlights the importance of variations in the exposure/resistance of infants and young children to infection in different settings, especially as this impacts on design of control programs; it predicts that naturally induced clinical and sterile immunity to typhoid is long lived and highlights the importance of the carrier state especially in areas of low transmission.
Collapse
Affiliation(s)
- Allan Saul
- Novartis Vaccines Institute for Global Health, Siena, Italy
- * E-mail:
| | - Tom Smith
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Nicolas Maire
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| |
Collapse
|
44
|
Abstract
Low-income countries typically lag behind industrialised nations, where the introduction of new vaccines is commonly tailored to the pressures of the commercial market. Happily in recent years this paradigm has started to change with the introduction of a univalent meningococcal A conjugate vaccine that is specifically targeted for the prevention of epidemic meningitis in Africa. The declaration of the 2010s as a New Decade of Vaccines, together with Millennium Development Goals 4 and 5, provide a strong mandate for a new approach to the development of vaccines for low-income countries, so that there has never been a more exciting time to work in this field. This review considers the opportunities and challenges of developing these new vaccines in the context of innovations in vaccinology, the need to induce protective immunity in the populations at risk and the requirement for strong partnership between the countries that will use these vaccines and different elements of the vaccine industry.
Collapse
|
45
|
A human IgG anti-Vi reference for Salmonella typhi with weight-based antibody units assigned. Vaccine 2013; 31:1970-4. [PMID: 23422143 DOI: 10.1016/j.vaccine.2013.02.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 01/08/2013] [Accepted: 02/04/2013] [Indexed: 11/22/2022]
Abstract
Recent data showing the high incidence of typhoid fever in young children, the demonstration of safety and efficacy of a Vi conjugate for this age group, the safety and similar immunogenicity in infants when administrated concurrently with EPI vaccines, together with the interests of manufacturers and investigators in studying such conjugate vaccines prompted us to prepare a human IgG anti-Vi standard to facilitate this work. Volunteers were injected with an investigational Vi-recombinant Pseudomonas aeruginosa exoprotein A (Vi-rEPA) conjugate vaccine. Plasmas with the highest levels of IgG anti-Vi were pooled. The IgG anti-Vi content of this preparation, assayed by precipitin analysis with purified Vi, was 33 μg/ml. Accordingly, the estimated IgG anti-Vi protective level of 3.5 ELISA unit/ml, derived from our efficacy trial of Vi-rEPA in 2-5 years old children, is equivalent to 4.3 μg/ml. This reagent is suitable for comparison of immune response of Vi conjugate vaccines or for other purposes requiring anti-Vi measurement.
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW This review summarizes the recent advances in vaccination against Salmonella enterica serovar Typhi and highlights the data supporting the development of next generation vaccines to address paratyphoid fever and invasive nontyphoidal Salmonella (iNTS) disease. RECENT FINDINGS There has been increasing awareness of the disease burden caused by S. Typhi particularly in Africa and greater recognition of S. Paratyphi A's contribution to enteric fever episodes throughout Asia. Groups have been working to improve the existing typhoid vaccines and provide comprehensive data on the feasibility of their implementation in endemic settings. These data have resulted in modifications to the recommendations for typhoid vaccination in traveller markets and endemic settings, and has also led to the development of S. Paratyphi A vaccine components that can be combined with existing typhoid vaccines to generate bivalent formulations against enteric fever. The epidemiology of iNTS serovars as cause of appreciable morbidity and mortality in Africa, and the need for vaccines, has also become more widely appreciated. SUMMARY Current typhoid vaccines, although moderately effective for short periods of time, cannot be used in all age groups and only target one of the clinically relevant Salmonella serovars. Greater effort must be placed on the development and implementation of improved vaccines for the disease burden resulting from Typhi, Paratyphi A or iNTS infections.
Collapse
|
47
|
Micoli F, Rondini S, Gavini M, Lanzilao L, Medaglini D, Saul A, Martin LB. O:2-CRM(197) conjugates against Salmonella Paratyphi A. PLoS One 2012; 7:e47039. [PMID: 23144798 PMCID: PMC3492368 DOI: 10.1371/journal.pone.0047039] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 09/07/2012] [Indexed: 11/18/2022] Open
Abstract
Enteric fevers remain a common and serious disease, affecting mainly children and adolescents in developing countries. Salmonella enterica serovar Typhi was believed to cause most enteric fever episodes, but several recent reports have shown an increasing incidence of S. Paratyphi A, encouraging the development of a bivalent vaccine to protect against both serovars, especially considering that at present there is no vaccine against S. Paratyphi A. The O-specific polysaccharide (O:2) of S. Paratyphi A is a protective antigen and clinical data have previously demonstrated the potential of using O:2 conjugate vaccines. Here we describe a new conjugation chemistry to link O:2 and the carrier protein CRM(197), using the terminus 3-deoxy-D-manno-octulosonic acid (KDO), thus leaving the O:2 chain unmodified. The new conjugates were tested in mice and compared with other O:2-antigen conjugates, synthesized adopting previously described methods that use CRM(197) as carrier protein. The newly developed conjugation chemistry yielded immunogenic conjugates with strong serum bactericidal activity against S. Paratyphi A.
Collapse
|
48
|
|
49
|
Immunization with the conjugate vaccine Vi-CRM₁₉₇ against Salmonella typhi induces Vi-specific mucosal and systemic immune responses in mice. Vaccine 2012; 30:6111-4. [PMID: 22705173 DOI: 10.1016/j.vaccine.2012.05.081] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 05/19/2012] [Accepted: 05/30/2012] [Indexed: 11/21/2022]
Abstract
Typhoid fever is a public health problem, especially among young children in developing countries. To address this need, a glycoconjugate vaccine Vi-CRM₁₉₇, composed of the polysaccharide antigen Vi covalently conjugated to the non-toxic mutant of diphtheria toxin CRM₁₉₇, is under development. Here, we assessed the antibody and cellular responses, both local and systemic, following subcutaneous injection of Vi-CRM₁₉₇. The glycoconjugate elicited Vi-specific serum IgG titers significantly higher than unconjugated Vi, with prevalence of IgG1 that persisted for at least 60 days after immunization. Vi-specific IgG, but not IgA, were present in intestinal washes. Lymphocytes proliferation after restimulation with Vi-CRM₁₉₇ was observed in spleen and mesenteric lymph nodes. These data confirm the immunogenicity of Vi-CRM₁₉₇ and demonstrate that the vaccine-specific antibody and cellular immune responses are present also in the intestinal tract, thus strengthening the suitability of Vi-CRM₁₉₇ as a promising candidate vaccine against Salmonella Typhi.
Collapse
|
50
|
Holmgren J, Svennerholm AM. Vaccines against mucosal infections. Curr Opin Immunol 2012; 24:343-53. [PMID: 22580196 DOI: 10.1016/j.coi.2012.03.014] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Accepted: 03/27/2012] [Indexed: 01/05/2023]
Abstract
There remains a great need to develop vaccines against many of the pathogens that infect mucosal tissues or have a mucosal port of entry. Parenteral vaccination may protect in some instances, but usually a mucosal vaccination route is necessary. Mucosal vaccines also have logistic advantages over injectable vaccines by being easier to administer, having less risk of transmitting infections and potentially being easier to manufacture. Still, however, only relatively few vaccines for human use are available: oral vaccines against cholera, typhoid, polio, and rotavirus, and a nasal vaccine against influenza. For polio, typhoid and influenza, in which the pathogens reach the blood stream, there is also an injectable vaccine alternative. A problem with available oral live vaccines is their reduced immunogenicity when used in developing countries; for instance, the efficacy of rotavirus vaccines correlates closely with the national per capita income. Research is needed to define the impact of factors such as malnutrition, aberrant intestinal microflora, concomitant infections, and preexisting immunity as well as of host genetic factors on the immunogenicity of these vaccines.
Collapse
Affiliation(s)
- Jan Holmgren
- University of Gothenburg Vaccine Research Institute (GUVAX) & Department of Microbiology and Immunology, The Sahlgrenska Academy at University of Gothenburg, Sweden.
| | | |
Collapse
|